• No results found

Short-Chain Fatty Acids Regulate Cytokines and Th17/Treg Cells in Human Peripheral Blood Mononuclear Cells in vitro

N/A
N/A
Protected

Academic year: 2020

Share "Short-Chain Fatty Acids Regulate Cytokines and Th17/Treg Cells in Human Peripheral Blood Mononuclear Cells in vitro"

Copied!
37
0
0

Loading.... (view fulltext now)

Full text

(1)

1

Short-Chain Fatty Acids Regulate Cytokines and Th17/Treg Cells in Human 1

Peripheral Blood Mononuclear Cells in vitro 2

3

4

Authors: 5

M. Asarata c, V. Apostolopoulosa b, T. Vasiljevica O. Donkor a* 6

aAdvanced Food Systems Research Unit, College of Health and Biomedicine, Victoria 7

University, Werribee Campus, P.O. Box 14428, Melbourne, Vic 8001, Australia. 8

bVAConsulting Services, P.O. Box 6437, Melbourne, Vic, 3030, Australia. 9

cFaculty of Veterinary Medicine, Tripoli University P.O. Box 13662, Tripoli, Libya. 10

*Corresponding author: Dr O. N. Donkor; Tel: (+61-3)-9919-8059, Fax: (+61-3)-99198284 11

e-mail: Osaana.Donkor@vu.edu.au 12

(2)

2 Abstract

14

Short chain fatty acids (SCFAs) have been recognized as mediators of immune responses, 15

including pathways of cytokine production. In this study, we investigated the immune-16

regulatory effects of SCFAs on human peripheral blood mononuclear cells (PBMCs) from 17

buffy coat of healthy donors. PBMCs were exposed to varying concentrations of individual 18

SCFAs or their mixtures, and, the production of interleukin (IL) IL-1β, 2, 6, 10, IL-19

17 IL-21 IL-23 and transforming growth factor beta 1 (TGF-β1) were assessed. T cell 20

differentiation after exposure to SCFAs was also examined. In comparison to LPS-stimulated 21

cells (controls), SCFAs slightly decreased the production of TGF-β1 and significantly 22

reduced IL-6 production (p < 0.05) and butyrate was more effective than acetate or 23

propionate; non-stimulated cells did not respond to SCFAs. In addition, the viability of 24

PBMCs was not significantly affected. SCFAs particularly butyrate caused the induction of 25

CD4+CD25+ regulatory T cells (Treg) rather than Th17 cells. It is clear that. SCFAs may up-26

regulate the production of anti-inflammatory cytokines in PBMCs resulting in the induction 27

of CD4+CD25+ Treg cells. 28

Key words: Short chain fatty acids, Human peripheral blood mononuclear cells, Cytokines, 29

Immune modulation, T helper cells, Th17 and CD4+CD25+ Treg cells. 30

(3)

3 1. Introduction

32

Short chain fatty acids (SCFAs) are a sub-group of fatty acids with 2 to 6 carbon atoms (C2 - 33

C6 mono-carboxylic acids), formed principally from fermentation of prebiotics by anaerobic 34

micro-organisms in the colon (Cummings & Macfarlane, 1991). SCFAs are predominant 35

anions in human colon contents (Sellin, 1999). Relative proportions and amounts of SCFAs 36

vary according to the type of fibre in the diet, type and population of microflora present in the 37

colon and the gut transit time, absorption and utilization by colonic epithelium (Macfarlane & 38

Macfarlane, 2003). Hence, the concentrations of SCFAs in the colon, portal blood and 39

peripheral circulation are different. Total SCFAs in the colon has been estimated to range 40

from 70 - 140 mM with concentration ratios of acetate, propionate and butyrate being, 10, 1.5 41

and 2 mM, respectively (Bergman, 1990; Cummings, Pomare, Branch, Naylor, & 42

Macfarlane, 1987; Sellin, 1999; Topping & Clifton, 2001). Whereas in the portal blood, 43

concentrations are typically higher (0.375 mM) compared to that in peripheral circulation 44

(0.079 mM) (Cummings et al., 1987). Likewise, Meijer, de Vos, and Priebe (2010) reported 45

the total concentration of SCFAs in human peripheral blood to ranged from 0.050 – 0.1 mM 46

and in portal blood, about 0.3 – 0.450 mM. 47

SCFAs have been shown to have beneficial effect on human health; they are the favoured 48

fuel of the colonic epithelium and are vital for intestinal and epithelial barrier functions 49

(Adom & Nie, 2013; Scheppach, 1994). Lack of source of energy may lead to diminished 50

integrity of epithelial function resulting in bowel disorders such as bowel inflammation 51

(Harig, Soergel, Komorowski, & Wood, 1989; Wong & Jenkins, 2007). Furthermore, SCFAs 52

stimulate colonic blood flow and enhance fluid and electrolytes uptake such as calcium 53

absorption (Roy, Kien, Bouthillier, & Levy, 2006). Recent interest in SCFAs benefits have 54

focused on their regulatory effects on immune responses by affecting the immune cells 55

(4)

4

pathological conditions such as inflammatory bowel disease (IBD), possibly via their effect 57

on immune responses (Tedelind, Westberg, Kjerrulf, & Vidal, 2007; Vernia et al., 1995; 58

Vinolo et al., 2011). However, the mechanism of action is not clearly demonstrated but it has 59

been reported that SCFAs may trigger cellular receptors such as G protein coupled receptors 60

(GPCRs), (GPR41 and GPR43), of immune cells and subsequently initiate the immune 61

response (Bindels, Dewulf, & Delzenne, 2013; Brown et al., 2003; Le Poul et al., 2003; 62

Maslowski et al., 2009; Masui et al., 2013). Moreover, studies have suggested that butyrate 63

and other SCFAs have inhibitory effects on the nuclear factor kappa B (NF-κB) signalling 64

(Liu et al., 2012; Luhrs et al., 2001; Segain et al., 2000; Tedelind et al., 2007) and histone 65

deacetylase (HDAC) (Aoyama, Kotani, & Usami, 2010; Waldecker, Kautenburger, 66

Daumann, Busch, & Schrenk, 2008). SCFAs may regulate cytokine production (Cavaglieri et 67

al., 2003; Masui et al., 2013; Yin, Laevsky, & Giardina, 2001), and, recently, butyrate and 68

propionate have been noted to promote peripheral regulatory T cell (Treg)differentiation 69

which might contribute to immune homeostasis (Arpaia et al., 2013). 70

PBMCs compose mainly of lymphocytes, macrophages and monocytes. The lymphocyte 71

population in healthy human adults consists of approximately 60% T cells (CD4+ and CD8+) 72

and 35% B cells, natural killer cells (NK), macrophage and monocytes (Minoprio, 2000; 73

Plebanski, 2002). PBMCs have been widely used in immunological and pharmaceutical 74

studies probably due to their properties, which relate to sophisticated immune cells. The cells 75

are co-cultured with different types of immune suppressant or stimulant drugs to study and 76

determine their efficiencies using different parameters of immune responses such as the 77

release of immune mediators (cytokines) (Ramachandran et al., 2012). T lymphocytes can be 78

functionally distinguished into cytotoxic (Tc) and helper (Th) cells. Human naive CD4+ T 79

helper cells can be divided into different functional subsets Th1, Th2, Th17, and Treg cells 80

(5)

5

2012). Th1 cells express T-box transcription factor (Tbet-2) and release IL2, TNFα and IFNγ 82

cytokines and act against intracellular pathogens. Th2 express GATA3, produce anti-83

inflammatory cytokines such as IL-1 receptor antagonist (IL-1ra), IL-4, IL-5, IL-6, IL-10 and 84

IL-13, and stimulate humoral immune responses against extracellular pathogens. Th17 cells 85

express the up-regulation of transcription factors, such as RORγt and STAT-3, release pro-86

inflammatory cytokines IL-17 and play a role in organ-specific autoimmune diseases 87

(Thomas Korn, Oukka, Kuchroo, & Bettelli, 2007). Treg cells express FoxP3 and produce IL-88

10 and TGF-β cytokines, promote tolerance to self and non-pathogenic antigens, suppress 89

amplitude immune and inflammatory responses, drive and modulate immune responses, and 90

abrogate autoimmune diseases (Wan & Flavell, 2009). 91

Mature CD4+ Th cells are also involved in autoimmunity, during T cell receptor (TCR) 92

activation in a particular cytokine milieu, naive CD4 T cells may differentiate into one of 93

several lineages of Th cells, including Th1, Th2, Th17, and induce Treg cells (iTreg) as 94

defined by their pattern of cytokine production and function. Imbalance of either pattern of 95

Th1 or Th2 cells may cause autoimmune disease e.g. a dysregulation of T helper cell 96

phenotype in favour of Th1 appears to underlie Crohn’s disease, a form of IBD (Dionne, 97

Ruemmele, & Seidman, 2004). Along with Th1 and Th2 cells, (FoxP3) Treg and Th17 cells 98

have been recognized as significant players in immune balance (Brand, 2009). The effect of 99

SCFAs on Treg cells may subsequently regulate the induction of Th1, Th2 and Th17 and 100

maintaining the immune homeostasis, thus SCFAs may be valuable to maintain the immune 101

homeostasis and prevent of chronic inflammation (Bailon et al., 2010; Vinolo et al., 2011). 102

SCFAs particularly butyrate have been known to modulate immune responses (Cox et al., 103

2009; Vinolo et al., 2011). However, little is known about the effects of SCFAs on human 104

PBMCs. Herein we evaluated the regulatory effects of SCFAs in PBMCs and the induction of 105

(6)

6

inflammatory cytokines with PBMCs in the presence or absence of LPS stimulation was also 107

examined. 108

2. Material and methods 109

2.1.Chemicals and reagent 110

Biological grade acetate, propionate, butyrate,purified LPS from Escherichia coli O111:B4 111

and growth medium Roswell Park Memorial Institute medium (RPMI-1640) were purchased 112

from Sigma (Sigma-Aldrich, Sydney, Australia). Ficoll-Paque™ Plus was from GE 113

Healthcare (GE Healthcare, Bio-Sciences, Uppsala, Sweden). Antibiotic-Antimycotic 114

solution and Fetal bovine serum (FBS) were acquired from Gibco Life Technologies 115

(Gibco®Life Technologies, Mulgrave, Australia). Phosphate-Buffered Saline (1X) pH 7.4, 116

(PBS) was from Invitrogen (Invitrogen Pty Ltd., Mount Waverly, Victoria, Australia),The 117

Buffy coat was provided by the Australian Red Cross Blood Services, Melbourne, Australia, 118

Other reagents and chemicals were of biotechnological and molecular-biology grade from 119

Sigma-Aldrich unless otherwise stated. 120

2.2.Isolation of human PBMCs from buffy coat using Ficoll gradient 121

In order to meet the requirements of the National Health and Medical Research Council 122

“National Statement on Ethical Conduct in Human Research” (National Health and Medical 123

Research Council, 2007), the Human research ethics of the proposed project was accepted 124

and approved by the Chair of the Faculty of Health, Engineering, and Science, Victoria 125

University Human Research Ethics Committee. A contract agreement was also conducted 126

with Australian Red Cross Blood Services Melbourne Australia, in order to the supply of 127

(7)

7

PBMCs were isolated from human buffy coat by Ficoll-Paque gradient according to the 129

method described by (Donkor et al., 2012) with minor modifications. Briefly, individual 130

buffy coat (60 mL) was diluted with an equal volume of phosphate buffer saline (PBS) and 131

layered on Ficoll-Paque Plus (GE Healthcare, Bio-Sciences, Uppsala, Sweden). Cells at the 132

interphase were collected following centrifugation (680 g, 25 min, 18°C) (Sorvall® RT7 133

centrifuge; DuPont, Newtown, CT, USA). Separated layers PBMC were washed twice in cold 134

PBS, and following centrifugation (680 g, 10 min, 18°C). To lyse any remaining red blood 135

cells, the pellet was resuspended in 5 mL red blood cell lysing buffer; Ammonium-Chloride-136

Potassium (ACK) (Gibco® ACK Life Technologies) and incubated for 8 min at room 137

temperature. The volume was then adjusted to 35 mL using sterile PBS then centrifuged 138

(680 g, 10 min, 18°C). Following two subsequent washes, the cell pellet was resuspended in 139

RPMI1640 medium supplemented with 10% FBS and 1% of Antibiotic-Antimycotic solution 140

for co-culture and stimulation. 141

2.3.Co-culture and stimulation of PBMCs by SCFAs

142

Human PBMCs were seeded in flat bottom 6-well tissue culture plates (Corning, Sigma) at 143

final concentration 1 × 106 cells/mL RPMI1640 medium per well, either in RPMI1640 144

medium alone or with LPS (controls), medium with LPS and acetate, propionate, butyrate or 145

mixed SCFAs. The concentrations of SCFAs were 1, 1.5, 2 mM similar to physiological 146

concentration found in the colon and peripheral circulation (portal vein) (Cummings et al., 147

1987; Topping & Clifton, 2001), similar concentrations were also used in previous studies 148

(Liu et al., 2012; Nancey et al., 2002; Weber & Kerr, 2006). LPS (5 μg/mL) was used for 149

cells stimulation (Chen, Bruns, Donnelly, & Wunderink, 2010; Jansky, Reymanova, & 150

(8)

8

To investigate the responses of PBMCs to the stimuli (LPS and/or SCFAs), the cells were 152

stimulated with 5µg/mL LPS alone (Chen et al., 2010), with SCFAs only or a combination of 153

LPS and SCFAs for 48 h. PBMCs were also stimulated with LPS for 24 hour then SCFAs 154

were added and cells further incubated for another 24 hours. 155

2.4.PBMCs viability assay 156

PBMCs viability in presence of LPS and/or SCFAs were assessed using MTS (3-(4,5-157

dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2- (4-sulfophenyl)-2H-tetrazolium, inner 158

salt) assay (CellTiter 96® AQueous One Solution Cell Proliferation Assay, Promega, USA) 159

according to manufacture instruction. Briefly, cells were seeded in 96-well plates (Corning, 160

Sigma) at concentration 1 × 103 cells per 100 µL of growth media with or without various 161

stimuli (as above), and then incubated for 48 h at 37°C in a 5% CO2 incubator. The viability 162

was detected by adding 20 µL of MTS solution, followed by 4 h of incubation then 163

absorbance was read at 490 nm using iMark Microplate Absorbance Reader (BIO-RAD, 164

Australia). After subtracting of the background reading, cell viability was calculated as: 165

% viable cell = (Optical density (OD) of SCFAs treated sample / OD of control sample) x 166

100. 167

2.5.ELISA analysis of cytokines 168

Supernatants from stimulated and non-stimulated PBMCs cultures were collected and 169

analysed for cytokines concentrations using BD OptEIA ELISA kits (BD Bioscience, San 170

Diego, CA), including IL-1β, IL-2, IL-6, IL-10, IL-17, IL-21 TGF-β1 and IL-23. The 171

detection procedures were performed in accordance with the manufacturer’s instructions. 172

(9)

9

cytokine response minus background (pg/ml) of each treatment from triplicate wells, plus or 174

minus the standard error of the mean. 175

2.6.Measurement of nitric oxide production 176

Nitric oxide (NO) production from stimulated PBMCs and non-stimulated PBMCs was 177

measured spectrophotometrically using Griess Reaction Assay (Promiga, Auburn, Australia) 178

according to the manufacture’s instruction. Briefly, after co-culture and stimulation of 179

PBMCs (as above), 50 µL aliquots of supernatant were incubated with 100 µL Griess reagent 180

(50 µL of 1% sulfanilamide in 0.1 M HCl and 50 µL of 0.1% N-1-napthylethylenediamine 181

dihydrochloride) for 10 min at room temperature. Absorbance was read at 550 nm using 182

iMark Microplate Absorbance Reader (BIO-RAD, Australia) and results were calculated 183

based on a NaNO2 standard curve. 184

2.7.Flow Cytometry Analysis of Th17 and Treg cells 185

Activated PBMCs were collected and analysed for induction of Treg/Th17 cells. Briefly, 1 186

mL suspension PBMCs culture was collected and centrifuged at 500g for 10 min, cell pellet 187

was washed twice using fluorescence activated cell sorting (FACS) buffer (PBS + 2% FBS) 188

and the suspension was centrifuged again at 500g for 10 min. PBMCs were re-suspended at 1 189

× 106 cells/ml in FACS buffer and surface marker staining was performed using fluorescein 190

isothiocynate (FITC)-labelled anti-human CD4, allophycocyanin labelled anti-human 191

CD25/CD3 (Becton-Dickinson), peridinin chlorophyll protein (PerCP)-labelled anti-human 192

CD3 (Biolegend, San Diego, CA, USA) and PerCP cyanine (Cy)5.5-labelled anti-human 193

CCR6 (CD196). Intracellular staining was performed using phycoerythrin (PE)-labelled anti-194

human FoxP3/RORγt (BD Pharmingen and R&D Systems, Minneapolis, MN, USA, 195

respectively), according to the manufacturer’s instructions. Samples were read using a BD 196

(10)

10

Biosciences). Analysis was performed using Gatelogic version 3.07 software (Inivai, 198

Victoria, Australia). Absolute numbers of Treg and Th17 cells were calculated as a 199

percentage of the total lymphocyte number (Donkor et al., 2012). 200

2.8.Statistical analysis 201

The results obtained were analysed as a split plot in time design with three main factors: 202

SCFAs (Three fatty acids) and their doses (3 dose levels) as the main plot and time (two time 203

frame 24 and 48) LPS was used as stimulant. The statistical evaluations of the data were 204

performed using the general linear model (GLM) (SAS/STAT., 1996). Significant differences 205

between treatments were tested by analysis of variance (ANOVA) followed by a comparison 206

between treatments performed by Fisher’s least significant difference (LSD) between each 207

sample with a level of significance of (p < 0.05). Data are expressed as mean and standard 208

deviation of triplicate measures determined in 5 independent experiments. 209

3. Results 210

3.1.PBMCs viability, proliferation and NO production 211

The MTS assay of PBMC viability and proliferation with or without different stimuli is 212

shown in Figure 1. Proliferation of PBMC were potentiated in the presence of LPS in all 213

samples compared with non LPS-stimulated PBMCs. Relatively similar effects resulted in the 214

presence of each SCFA with maximum proliferation observed in the presence of butyrate 215

followed by mixed SCFAs over 48 h.Similar LPS effect on PBMCs proliferation was 216

reported (Jansky et al., 2003), and SCFAs did not induce cell apoptosis at used concentration. 217

Production of NO was enhanced in the presence of LPS, whereas the addition of SCFAs 218

induced remarkable reduction (p < 0.05) of NO (Fig. 2). After 48 h of incubation, NO was 219

(11)

11

with LPS-stimulated cells, However the inhibition of NO in the SCFA mixture was similar to 221

that of butyrate. 222

3.2.Production of cytokines 223

Figure 3 (A-H) show cytokines (IL-β1, IL-2, IL-6, IL-17, IL-21 IL-23 and TGF-β1) produced 224

by PBMCs in the presence of LPS compared with non-stimulated PBMCs, the combination 225

of LPS and SCFAs or SCFAs only. Results show significant (p < 0.05) decrease in IL-β1, IL-226

2, IL-6, IL-17 and IL-21, with a slight reduction of IL-23 and TGF-β1 in the presence of 227

SCFAs, compared to cells stimulated with LPS only. However, IL-10 secretion was not 228

affected by LPS compared to non LPS-stimulated cells but it was increased in the presence 229

of SCFAs particularly in butyrate (Säemann et al., 2000). We have also noted similar results 230

when PBMCs stimulated with LPS for 24 h followed by addition of SCFAs and incubated for 231

another 24 hours (Table 1). 232

3.3.CD4+ CD25; Th17 and Treg populations 233

Flow cytometric analysis of LPS and SCFAs stimulated PBMCs showed increased CD4+ 234

CD25+ Treg and Th17 cells compared to unstimulated cells (Fig. 4). LPS aloneincreased the 235

Th17 cell population compared to Treg cells after 48 h of incubation. In the presence of 236

SCFAs particularly butyrate, the relative proportion of Th17 and Treg showed slight increase 237

of Treg cells compared to Th17 (Fig. 4). The addition of SCFAs along with LPS at 0 time or 238

after the pre-stimulated PBMCs with LPS, showed comparatively similar up-regulation of 239

Treg cells after 24 h compared to unstimulated cells (control). Butyrate was more effective in 240

induction of Treg cells than acetate or propionate. The addition of mixed SCFAs did not 241

induce augmentation of the effects as compared with each SCFA. However, their effects 242

were concentration-dependent; 2 mM of SCFAs was more effective in up-regulation of Treg 243

(12)

12 4. Discussion

245

Despite most of intestinal SCFAs being used by intestinal epithelium cells, a considerable 246

amount of acetate, propionate, and butyrate are absorbed into the blood and exert their effects 247

at peripheral tissue level beyond the digestive system through regulation of immune 248

responses (Matsumoto et al., 2006). Consequently, we assumed that SCFAs might have 249

regulatory effects on peripheral PBMCs. Therefore, in the current study, we used in vitro 250

LPS-stimulated PBMCs of healthy donor to compare the anti-inflammatory effect of acetate, 251

propionate, and butyrate and regulation of Th17 and Treg balance. It was found that SCFAs 252

reduced the production of pro-inflammatory factors including IL-1β, IL-6, IL-17 and NO 253

whereas they enhanced the production of anti-inflammatory mediators such as 10, and IL-254

2. Furthermore, SCFAs affected gene expression of T helper cells possibly through their 255

effect on immune mediators and growth factors such as IL-6 and TGF-β1, which might have 256

led to the up-regulation of Treg cells. Interestingly, SCFAs exhibit these effects mainly in 257

LPS-stimulated PBMCs whereas non LPS-stimulated cells were not affected by SCFAs. 258

4.1.Effect of SCFAs on cells viability 259

To determine whether SCFAs exert their effects through stimulation of PBMCs but not via 260

induce cellular death, MTS assay was conducted after co-culturing of PBMCs with LPS and 261

SCFAs (Fig. 1). Cell growth was slightly potentiated in the presence of LPS and SCFAs 262

particularly butyrate or mixed SCFAs, indicating that viability of cells was not affected by 263

experimental conditions. Furthermore, slight proliferation of PBMCs following LPS exposure 264

in all samples indicated that the proliferation was mainly due to stimulation effect of LPS and 265

that the SCFAs only promoted the LPS activated cell proliferation. The induction of cell 266

proliferation indicate that LPS was able to trigger cell response and initiate the immune 267

(13)

13

cells, monocytes, and macrophages. This was expected as LPS is known to induce a 269

macrophage-dependent immune response through activation of NF-κB transcription factor, 270

subsequently enhancing proliferation and release of immune factors from PBMCs (Martich, 271

Boujoukos, & Suffredini, 1993; Sharif, Bolshakov, Raines, Newham, & Perkins, 2007). In 272

our study, acetate, propionate and butyrate did not cause apoptosis in PBMCs but promoted 273

the proliferation of LPS-stimulated PBMCs. Different effects of SCFAs on proliferation of 274

PBMCs and other similar cells have been highlighted (Meijer et al., 2010). A study on the 275

effect of SCFAs in mouse macrophage cell line RAW264.7 reported that viability of cells 276

was not affected when incubated with 0 - 1.2 mM SCFAs (Liu et al., 2012). Similarly, an 277

earlier study demonstrated that unstimulated PBMCs were not affected by SCFAs at a 278

physiological level (Cox et al., 2009). On the other hand some studies showed that butyrate 279

caused apoptosis in antigen stimulated T cells and macrophages (Bailon et al., 2010; Kurita-280

Ochiai, Fukushima, & Ochiai, 1999). For example, 2 mM of butyric acid induced inhibition 281

of proliferation after concanavalin-A stimulated porcine PBMCs (Weber & Kerr, 2006). 282

Another study revealed that lymphocytes proliferation was inhibited due to 1.5 mM of 283

butyrate but not acetate or propionate at the same concentration (Cavaglieri et al., 2003). 284

These different effects of SCFAs might be due to the using of different concentrations, 285

sources of PBMCs and method of stimulation (Meijer et al., 2010). 286

4.2.Effect of SCFAs on cytokine release 287

Lymphocytes besides inflammatory cells are involved in immune and inflammatory 288

responses. They interact with each other via release of cytokines and expression of cytokine 289

receptors in response to stimuli (Gruys, Toussaint, Niewold, & Koopmans, 2005). Cytokines 290

can act as positive or negative regulators of immune responses and maintain lymphocyte 291

balance. Nevertheless, the activity of cytokines are dependent upon each other along with 292

(14)

14

Schmidt-Arras, & Rose-John, 2011). Factors affecting cytokine network may subsequently 294

influence the inflammatory or immune response. Among these factors, SCFAs which have 295

been recognized as regulators of immune response through different mechanisms, such as 296

directly affecting the immune cells by binding to specific receptors GPCR (Le Poul, et al., 297

2003), inhibition of HDAC (Davie, 2003), suppression of activation of the transcription 298

factor NF-κB (Segain et al., 2000) and revelling different anti-inflammatory activities via 299

regulation of cytokines secretion by immune cells (Cox et al., 2009; Saulnier, Spinler, 300

Gibson, & Versalovic, 2009). 301

The current study highlights the effect of SCFAs on secretion of cytokines that are involved 302

in T cell differentiation into either Th17 or Treg cells besides the main cytokines that are 303

released by Th17 and Treg. Stimulation of PBMCs with LPS alone lead to an increased 304

production of IL-6 and TGF-β as LPS is a known stimulant to induce the secretion of pro-305

inflammatory cytokines (Jansky et al., 2003; Martich et al., 1993; Sharif et al., 2007). The 306

combination of LPS and SCFAs particularly butyrate decreased IL-6 and marginally reduced 307

TGF-β1. These effects were similar for either SCFAs added to 24 h pre-stimulated PBMCs or 308

at 0 time of stimulation (Table 1 and Fig. 3). The reduction of IL-6 and slightly reduced TGF-309

β1concentration in the pre-stimulated PBMCs was probably due to the neutralizing and 310

inhibiting effects of SCFAs on pro-inflammatory condition in stimulated PBMCs. This might 311

have mediated the secretion of anti-inflammatory cytokines that likely act as immune 312

regulator of pro-inflammatory cytokines (Dinarello, 1997; Opal & DePalo, 2000; Sultani, 313

Stringer, Bowen, & Gibson, 2012). The effect of SCFAs on immune cells depends on the 314

activation status and differentiation stages of effector cells (Cox et al., 2009). Furthermore, 315

incubation of PBMCs in the presence of SCFAs without LPS did not induce changes in the 316

release of tested cytokines an indication that the physiological concentrations of SCFAs had 317

(15)

15

TGF-β1 has verity of functions, the exposure of PBMCs to TGF-β1 can generate a variety of 319

cellular processes including inhibition of proliferation, differentiation, migration and 320

apoptosis (Sanchez-Capelo, 2005). TGF-β1 regulates many other growth factors and plays a 321

role in naive cell differentiation based on its concentration and other cytokine environment. 322

TGF-β1 in the presence of IL- 6 , IL-1β, IL-21 or IL-23 drive cell differentiation to Th17 323

cells, subsequently releasing more pro-inflammatory factors (Yoshimura, Suzuki, Sakaguchi, 324

Hanada, & Yasukawa, 2012; Liang Zhou, Chong, & Littman, 2009; Liang Zhou et al., 2007). 325

Little is known about effects of SCFAs on release of TGF-β1 in human PBMCs, in our study 326

important pleiotropic cytokine, TGF-β1 was increased significantly in the presence of LPS 327

but only slight decrease with SCFAs. Increase in the concentration of IL-6 along with TGF-328

β1 could trigger differentiation of CD4+ to Th17 cells. The induction of Th17 cells could be 329

related to the down-regulation of IL-6 but not TGF-β1 since the level of TGF-β1 was not 330

significantly affected by SCFAs. Consistent with our study SCFAs have been reported to 331

supress pro-inflammatory mediators such as TNF-α, IL-6 and enhance the release of anti-332

inflammatory cytokine IL-10 (Meijer et al., 2010; Park, Lee, Lee, Kim, & Kim, 2007). 333

Pleiotropic cytokines TGF-β1 plays important role in regulation of immune response by 334

acting with other cytokines such as IL-2 and IL-10 to promote expression and activation of 335

Treg cells, and released more IL-2 and IL-10, consequently up-regulating anti-inflammatory 336

condition and ameliorates inflammation (Taylor, Verhagen, Blaser, Akdis, & Akdis, 2006). 337

4.3.Differentiation of CD4+ cells 338

Based on the cytokine milieu, activated naïve T helper cells may be differentiated into Th1, 339

Th2, Th17 or Treg phenotypes with different effector roles and cytokine profiles (Broere, 340

Apasov, Sitkovsky, & van Eden, 2011). IL-12 milieu skews CD4+ T helper cells to Th1, IL-4 341

skews CD4+ T helper cells to Th2, TGF-β1 skews CD4+ T helper cells to Treg and IL-6 with 342

(16)

16

Kimura & Kishimoto, 2010; Ramgolam, Sha, Jin, Zhang, & Markovic-Plese, 2009). 344

Furthermore other cytokines also play a role in cell differentiation and activation such as IL-2 345

which act with TGF-β1 to drive CD4+ T cells to Treg cells (Campbell & Koch, 2011; Zheng, 346

2013; Ziegler & Buckner, 2009) and TGF-β with IL-1β, IL-21, and IL-23 are implicated in 347

promoting human Th17 differentiation (T. Korn, Bettelli, Oukka, & Kuchroo, 2009; 348

Ramgolam et al., 2009; Yang et al., 2008; Zheng, 2013; Ziegler & Buckner, 2009). In our 349

study the, down regulation of Th17 cells may due to decline of IL-21 and IL-23 rather than 350

reduction of TGF-β which was relatively stable in our study condition (Fig. 3). 351

Treg cells and Th17 cells are two subtypes of CD4+ cells. They play opposing roles in 352

autoimmune inflammatory diseases and immune tolerance, although they share a common 353

differentiation pathway. Imbalance of Treg and Th17 has been established in several 354

autoimmune diseases (Ji et al., 2012). Recent studies in human and mouse CD4+ T cells show 355

dichotomy in the generation and differentiation of Treg cells and Th17 cells (Kimura & 356

Kishimoto, 2010; Zhu, Yamane, & Paul, 2010). The TGF-β1 signalling is described as the 357

co-expression of Foxp3 and RORγ-t (L. Zhou et al., 2008). This signalling depends on other 358

immune factors. For example, TGF-β1 alone enhances Foxp3 expression and inhibits RORγ-t 359

activity, whereas combination of TGF-β1 with either IL-1β and IL-23 or IL-21 and IL-6 360

drives human Th17 differentiation (de Jong, Suddason, & Lord, 2010). This indicates the 361

inner correlation between Th17 and Treg cells through the cytokine milieu (Ji et al., 2012). 362

SCFAs are supposed to have a regulatory influence on inflammatory disorder and ameliorate 363

inflammation in some intestinal inflammatory disorders (Cox et al., 2009). This might be 364

mediated through modulation of cytokine milieu in the medium and expression of Treg and 365

Th17. Our finding exhibited that Foxp3 and RORγ-t expression in T lymphocytes subset 366

population resulted after incubation of LPS stimulated PBMCs with different SCFAs. The 367

(17)

17

leading to increased frequency of Th17 and the subsequent release of IL-17 and IL-21. 369

However in the presence of SCFAs, Th17 cell differentiation was likely supressed and 370

favoured cytokine environment for FoxP3 regulatory cell induction leading to enhanced 371

FoxP3 expression. Butyrate was more effective in this regard than acetate and propionate. 372

These findings are consistent with studies suggesting that butyrate showed strong anti-373

inflammatory properties (Cavaglieri et al., 2003; Liu et al., 2012; Meijer et al., 2010; 374

Tedelind et al., 2007) Furthermore, Arpaia et al. (2013) and Furusawa et al. (2013) reported 375

that butyrate promotes the regulation of intestinal Treg generation. However, the effect of 376

SCFAs on T cell phenotypes need more studies. 377

5. Conclusion 378

SCFAs may have in vitro anti-inflammatory and immune regulatory effects through induction 379

of Treg cells and production of anti-inflammatory cytokines. Butyrate showed more 380

regulatory effect than propionate and acetate respectively. Our finding indicates that, SCFAs 381

may have regulatory properties on inflammatory processes via the balance of Th17/Treg cells 382

and pro and anti-inflammatory cytokines. 383

Acknowledgements 384

We gratefully acknowledge the Ministry of High Education of the Libyan Government and 385

Victoria University for their financial and technical support. We thank the Australian Red 386

Cross Services for their supply of blood products and technical supports by Burnet Institute, 387

and we gratefully acknowledge researchers and technical staff at Werribee Campus, Victoria 388

(18)

18 Reference

390

Adom, D., & Nie, D. (2013). Regulation of Autophagy by Short Chain Fatty Acids in Colon 391

Cancer Cells. 392

Afzali, B., Lombardi, G., Lechler, R. I., & Lord, G. M. (2007). The role of T helper 17 393

(Th17) and regulatory T cells (Treg) in human organ transplantation and autoimmune 394

disease. Clin Exp Immunol, 148(1), 32-46. doi: 10.1111/j.1365-2249.2007.03356.x 395

Aoyama, M., Kotani, J., & Usami, M. (2010). Butyrate and propionate induced activated or 396

non-activated neutrophil apoptosis via HDAC inhibitor activity but without activating 397

GPR-41/GPR-43 pathways. Nutrition, 26(6), 653-661. doi: 398

http://dx.doi.org/10.1016/j.nut.2009.07.006 399

Arpaia, N., Campbell, C., Fan, X., Dikiy, S., van der Veeken, J., Liu, H., Rudensky, A. Y. 400

(2013). Metabolites produced by commensal bacteria promote peripheral regulatory 401

T-cell generation. Nature, 504(7480), 451-455. 402

Bailon, E., Cueto-Sola, M., Utrilla, P., Rodriguez-Cabezas, M. E., Garrido-Mesa, N., 403

Zarzuelo, A., Comalada, M. (2010). Butyrate in vitro immune-modulatory effects 404

might be mediated through a proliferation-related induction of apoptosis. 405

Immunobiology, 215(11), 863-873. doi: 10.1016/j.imbio.2010.01.001 406

Bergman, E. N. (1990). Energy contributions of volatile fatty acids from the gastrointestinal 407

tract in various species. Physiological Reviews, 70(2), 567-590. 408

Bindels, L. B., Dewulf, E. M., & Delzenne, N. M. (2013). GPR43/FFA2: physiopathological 409

relevance and therapeutic prospects. Trends in pharmacological sciences, 34(4), 226-410

232. 411

Brand, S. (2009). Crohn's disease: Th1, Th17 or both? The change of a paradigm: new 412

immunological and genetic insights implicate Th17 cells in the pathogenesis of 413

(19)

19

Broere, F., Apasov, S. G., Sitkovsky, M. V., & van Eden, W. (2011). A2 T cell subsets and T 415

cell-mediated immunity Principles of Immunopharmacology (pp. 15-27): Springer. 416

Brown, A. J., Goldsworthy, S. M., Barnes, A. A., Eilert, M. M., Tcheang, L., Daniels, D., 417

Dowell, S. J. (2003). The Orphan G Protein-coupled Receptors GPR41 and GPR43 418

Are Activated by Propionate and Other Short Chain Carboxylic Acids. Journal of 419

Biological Chemistry, 278(13), 11312-11319. doi: 10.1074/jbc.M211609200 420

Campbell, D. J., & Koch, M. A. (2011). Phenotypical and functional specialization of 421

FOXP3+ regulatory T cells. Nat Rev Immunol, 11(2), 119-130. doi: 10.1038/nri2916 422

Cavaglieri, C. R., Nishiyama, A., Fernandes, L. C., Curi, R., Miles, E. A., & Calder, P. C. 423

(2003). Differential effects of short-chain fatty acids on proliferation and production 424

of pro- and anti-inflammatory cytokines by cultured lymphocytes. Life Sciences, 425

73(13), 1683-1690. doi: http://dx.doi.org/10.1016/S0024-3205(03)00490-9 426

Chen, J., Bruns, A. H., Donnelly, H. K., & Wunderink, R. G. (2010). Comparative in vitro 427

stimulation with lipopolysaccharide to study TNFalpha gene expression in fresh 428

whole blood, fresh and frozen peripheral blood mononuclear cells. J Immunol 429

Methods, 357(1-2), 33-37. 430

Cox, M. A., Jackson, J., Stanton, M., Rojas-Triana, A., Bober, L., Laverty, M., Jenh, C. H. 431

(2009). Short-chain fatty acids act as antiinflammatory mediators by regulating 432

prostaglandin E(2) and cytokines. World J Gastroenterol, 15(44), 5549-5557. 433

Cummings, J. H., & Macfarlane, G. T. (1991). The control and consequences of bacterial 434

fermentation in the human colon. Journal of Applied Microbiology, 70(6), 443-459. 435

doi: 10.1111/j.1365-2672.1991.tb02739.x 436

Cummings, J. H., Pomare, E. W., Branch, W. J., Naylor, C. P., & Macfarlane, G. T. (1987). 437

Short chain fatty acids in human large intestine, portal, hepatic and venous blood. 438

(20)

20

Davie, J. R. (2003). Inhibition of Histone Deacetylase Activity by Butyrate. The Journal of 440

Nutrition, 133(7), 2485S-2493S. 441

de Jong, E., Suddason, T., & Lord, G. M. (2010). Translational mini-review series on Th17 442

cells: development of mouse and human T helper 17 cells. Clin Exp Immunol, 159(2), 443

148-158. doi: 10.1111/j.1365-2249.2009.04041.x 444

Dinarello, C. A. (1997). Proinflammatory and anti-inflammatory cytokines as mediators in 445

the pathogenesis of septic shock. CHEST Journal, 112(6_Supplement), 321S-329S. 446

Dionne, S., Ruemmele, F. M., & Seidman, E. G. (2004). Immunopathogenesis of 447

inflammatory bowel disease: role of cytokines and immune cell-enterocyte 448

interactions. 449

Donkor, O. N., Ravikumar, M., Proudfoot, O., Day, S. L., Apostolopoulos, V., Paukovics, G., 450

Gill, H. (2012). Cytokine profile and induction of T helper type 17 and regulatory T 451

cells by human peripheral mononuclear cells after microbial exposure. Clinical & 452

Experimental Immunology, 167(2), 282-295. doi: 10.1111/j.1365-2249.2011.04496.x 453

Gruys, E., Toussaint, M. J., Niewold, T. A., & Koopmans, S. J. (2005). Acute phase reaction 454

and acute phase proteins. J Zhejiang Univ Sci B, 6(11), 1045-1056. doi: 455

10.1631/jzus.2005.B1045 456

Harig, J. M., Soergel, K. H., Komorowski, R. A., & Wood, C. M. (1989). Treatment of 457

diversion colitis with short-chain-fatty acid irrigation. New England Journal of 458

Medicine, 320(1), 23-28. 459

Jansky, L., Reymanova, P., & Kopecky, J. (2003). Dynamics of cytokine production in 460

human peripheral blood mononuclear cells stimulated by LPS or infected by Borrelia. 461

(21)

21

Ji, L., Zhan, Y., Hua, F., Li, F., Zou, S., Wang, W., Cheng, Y. (2012). The Ratio of 463

Treg/Th17 Cells Correlates with the Disease Activity of Primary Immune 464

Thrombocytopenia. PLoS ONE, 7(12), e50909. doi: 10.1371/journal.pone.0050909 465

Kimura, A., & Kishimoto, T. (2010). IL-6: Regulator of Treg/Th17 balance. European 466

Journal of Immunology, 40(7), 1830-1835. doi: 10.1002/eji.201040391 467

Korn, T., Bettelli, E., Oukka, M., & Kuchroo, V. K. (2009). IL-17 and Th17 Cells Annual 468

Review of Immunology (Vol. 27, pp. 485-517). Palo Alto: Annual Reviews. 469

Korn, T., Oukka, M., Kuchroo, V., & Bettelli, E. (2007). Th17 cells: Effector T cells with 470

inflammatory properties. Seminars in Immunology, 19(6), 362-371. doi: 471

http://dx.doi.org/10.1016/j.smim.2007.10.007 472

Kurita-Ochiai, T., Fukushima, K., & Ochiai, K. (1999). Lipopolysaccharide Stimulates 473

Butyric Acid-Induced Apoptosis in Human Peripheral Blood Mononuclear Cells. 474

Infection and Immunity, 67(1), 22-29. 475

Le Poul, E., Loison, C., Struyf, S., Springael, J.-Y., Lannoy, V., Decobecq, M.-E., Detheux, 476

M. (2003). Functional Characterization of Human Receptors for Short Chain Fatty 477

Acids and Their Role in Polymorphonuclear Cell Activation. Journal of Biological 478

Chemistry, 278(28), 25481-25489. doi: 10.1074/jbc.M301403200 479

Liu, T., Li, J., Liu, Y., Xiao, N., Suo, H., Xie, K., Wu, C. (2012). Short-chain fatty acids 480

suppress lipopolysaccharide-induced production of nitric oxide and proinflammatory 481

cytokines through inhibition of NF-κB pathway in RAW264. 7 cells. Inflammation, 482

35(5), 1676-1684. 483

Luckheeram, R. V., Zhou, R., Verma, A. D., & Xia, B. (2012). CD4+T Cells: Differentiation 484

and Functions. Clinical and Developmental Immunology, 2012, 12. doi: 485

(22)

22

Luhrs, H., Gerke, T., Boxberger, F., Backhaus, K., Melcher, R., Scheppach, W., & Menzel, 487

T. (2001). Butyrate inhibits interleukin-1-mediated nuclear factor-kappa B activation 488

in human epithelial cells. [Research Support, Non-U S Gov't]. Dig Dis Sci, 46(9), 489

1968-1973. 490

Macfarlane, S., & Macfarlane, G. T. (2003). Regulation of short-chain fatty acid production. 491

Proceedings of the Nutrition Society, 62(01), 67-72. doi: doi:10.1079/PNS2002207 492

Martich, G. D., Boujoukos, A. J., & Suffredini, A. F. (1993). Response of man to endotoxin. 493

[Review]. Immunobiology, 187(3-5), 403-416. 494

Maslowski, K. M., Vieira, A. T., Ng, A., Kranich, J., Sierro, F., Di, Y., Mackay, C. R. (2009). 495

Regulation of inflammatory responses by gut microbiota and chemoattractant receptor 496

GPR43. [10.1038/nature08530]. Nature, 461(7268), 1282-1286. doi: 497

http://www.nature.com/nature/journal/v461/n7268/suppinfo/nature08530_S1.html 498

Masui, R., Sasaki, M., Funaki, Y., Ogasawara, N., Mizuno, M., Iida, A., Kasugai, K. (2013). 499

G protein-coupled receptor 43 moderates gut inflammation through cytokine 500

regulation from mononuclear cells. Inflamm Bowel Dis, 19(13), 2848-2856. doi: 501

10.1097/01.MIB.0000435444.14860.ea 502

Matsumoto, N., Riley, S., Fraser, D., Al-Assaf, S., Ishimura, E., Wolever, T., Phillips, A. O. 503

(2006). Butyrate modulates TGF-β1 generation and function: Potential renal benefit 504

for Acacia (sen) SUPERGUM™(gum arabic)? Kidney international, 69(2), 257-265. 505

Meijer, K., de Vos, P., & Priebe, M. G. (2010). Butyrate and other short-chain fatty acids as 506

modulators of immunity: what relevance for health? Curr Opin Clin Nutr Metab 507

Care, 13(6), 715-721. doi: 10.1097/MCO.0b013e32833eebe5 508

Minoprio, P. (2000). Lymphocytes: A Practical Approach (2nd edn) by S.L. Rowland-Jones 509

and A.J. McMichel. Parasitology Today, 16(8), 360. doi:

510

(23)

23

Nancey, S., Bienvenu, J., Coffin, B., Andre, F., Descos, L., & Flourie, B. (2002). Butyrate 512

strongly inhibits in vitro stimulated release of cytokines in blood. Dig Dis Sci, 47(4), 513

921-928. 514

Opal, S. M., & DePalo, V. A. (2000). Anti-inflammatory cytokines. CHEST Journal, 117(4), 515

1162-1172. 516

Park, J. S., Lee, E. J., Lee, J. C., Kim, W. K., & Kim, H. S. (2007). Anti-inflammatory effects 517

of short chain fatty acids in IFN-gamma-stimulated RAW 264.7 murine macrophage 518

cells: involvement of NF-kappaB and ERK signaling pathways. Int 519

Immunopharmacol, 7(1), 70-77. doi: 10.1016/j.intimp.2006.08.015 520

Plebanski, M. (2002). Preparation of Lymphocytes and Identification of Lymphocytes 521

Subpopulations. In S. L. R.-J. a. A. McMichael (Ed.), Lymphocytes A Practical 522

Approach. New York, USA: Oxford University Press. 523

Ramachandran, H., Laux, J., Moldovan, I., Caspell, R., Lehmann, P. V., & Subbramanian, R. 524

A. (2012). Optimal thawing of cryopreserved peripheral blood mononuclear cells for 525

use in high-throughput human immune monitoring studies. Cells, 1(3), 313-324. 526

Ramgolam, V. S., Sha, Y., Jin, J., Zhang, X., & Markovic-Plese, S. (2009). IFN-β Inhibits 527

Human Th17 Cell Differentiation. The Journal of Immunology, 183(8), 5418-5427. 528

doi: 10.4049/jimmunol.0803227 529

Roy, C. C., Kien, C. L., Bouthillier, L., & Levy, E. (2006). Short-chain fatty acids: ready for 530

prime time? Nutr Clin Pract, 21(4), 351-366. 531

Säemann, M. D., Böhmig, G. A., Österreicher, C. H., Burtscher, H., Parolini, O., Diakos, C., 532

Zlabinger, G. J. (2000). Anti-inflammatory effects of sodium butyrate on human 533

monocytes: potent inhibition of IL-12 and up-regulation of IL-10 production. The 534

(24)

24

Sanchez-Capelo, A. (2005). Dual role for TGF-beta1 in apoptosis. Cytokine Growth Factor 536

Rev, 16(1), 15-34. doi: 10.1016/j.cytogfr.2004.11.002 537

SAS/STAT. (1996). SAS/STAT® Software: Changes and Enhancements: Through Release 538

6.11: SAS Institute Cary, North Carolina. 539

Saulnier, D. M. A., Spinler, J. K., Gibson, G. R., & Versalovic, J. (2009). Mechanisms of 540

probiosis and prebiosis: considerations for enhanced functional foods. Current 541

Opinion in Biotechnology, 20(2), 135-141. doi: 10.1016/j.copbio.2009.01.002 542

Scheller, J., Chalaris, A., Schmidt-Arras, D., & Rose-John, S. (2011). The pro- and anti-543

inflammatory properties of the cytokine interleukin-6. Biochimica et Biophysica Acta 544

(BBA) - Molecular Cell Research, 1813(5), 878-888. doi: 545

http://dx.doi.org/10.1016/j.bbamcr.2011.01.034 546

Scheppach, W. (1994). Effects of short chain fatty acids on gut morphology and function. 547

Gut, 35(1 Suppl), S35-38. 548

Segain, J. P., Raingeard de la Bletiere, D., Bourreille, A., Leray, V., Gervois, N., Rosales, C., 549

Galmiche, J. P. (2000). Butyrate inhibits inflammatory responses through NFkappaB 550

inhibition: implications for Crohn's disease. Gut, 47(3), 397-403. 551

Sellin, J. H. (1999). SCFAs: The Enigma of Weak Electrolyte Transport in the Colon. News 552

Physiol Sci, 14, 58-64. 553

Sharif, O., Bolshakov, V. N., Raines, S., Newham, P., & Perkins, N. D. (2007). 554

Transcriptional profiling of the LPS induced NF-kappaB response in macrophages. 555

BMC Immunol, 8, 1. 556

Sultani, M., Stringer, A. M., Bowen, J. M., & Gibson, R. J. (2012). Anti-Inflammatory 557

Cytokines: Important Immunoregulatory Factors Contributing to Chemotherapy-558

Induced Gastrointestinal Mucositis. Chemotherapy Research and Practice, 2012, 11. 559

(25)

25

Taylor, A., Verhagen, J., Blaser, K., Akdis, M., & Akdis, C. A. (2006). Mechanisms of 561

immune suppression by interleukin-10 and transforming growth factor-beta: the role 562

of T regulatory cells. Immunology, 117(4), 433-442. doi: 10.1111/j.1365-563

2567.2006.02321.x 564

Tedelind, S., Westberg, F., Kjerrulf, M., & Vidal, A. (2007). Anti-inflammatory properties of 565

the short-chain fatty acids acetate and propionate: a study with relevance to 566

inflammatory bowel disease. World J Gastroenterol, 13(20), 2826-2832. 567

Topping, D. L., & Clifton, P. M. (2001). Short-Chain Fatty Acids and Human Colonic 568

Function: Roles of Resistant Starch and Nonstarch Polysaccharides. Physiological 569

Reviews, 81(3), 1031-1064. 570

Vernia, P., Marcheggiano, A., Caprilli, R., Frieri, G., Corrao, G., Valpiani, D., Torsoli, A. 571

(1995). Short-chain fatty acid topical treatment in distal ulcerative colitis. Aliment 572

Pharmacol Ther, 9(3), 309-313. 573

Vinolo, M. A., Rodrigues, H. G., Nachbar, R. T., & Curi, R. (2011). Regulation of 574

inflammation by short chain fatty acids. Nutrients, 3(10), 858-876. doi: 575

10.3390/nu3100858 576

Waldecker, M., Kautenburger, T., Daumann, H., Busch, C., & Schrenk, D. (2008). Inhibition 577

of histone-deacetylase activity by short-chain fatty acids and some polyphenol 578

metabolites formed in the colon. The Journal of Nutritional Biochemistry, 19(9), 587-579

593. doi: http://dx.doi.org/10.1016/j.jnutbio.2007.08.002 580

Wan, Y. Y., & Flavell, R. A. (2009). How diverse--CD4 effector T cells and their functions. J 581

Mol Cell Biol, 1(1), 20-36. 582

Weber, T. E., & Kerr, B. J. (2006). Butyrate differentially regulates cytokines and 583

proliferation in porcine peripheral blood mononuclear cells. Veterinary immunology 584

(26)

26

Wong, J. M. W., & Jenkins, D. J. A. (2007). Carbohydrate Digestibility and Metabolic 586

Effects. The Journal of Nutrition, 137(11), 2539S-2546S. 587

Yang, L., Anderson, D. E., Baecher-Allan, C., Hastings, W. D., Bettelli, E., Oukka, M., 588

Hafler, D. A. (2008). IL-21 and TGF-beta are required for differentiation of human 589

T(H)17 cells. Nature, 454(7202), 350-352. doi: 10.1038/nature07021 590

Yin, L., Laevsky, G., & Giardina, C. (2001). Butyrate Suppression of Colonocyte NF-κB 591

Activation and Cellular Proteasome Activity. Journal of Biological Chemistry, 592

276(48), 44641-44646. doi: 10.1074/jbc.M105170200 593

Yoshimura, A., Suzuki, M., Sakaguchi, R., Hanada, T., & Yasukawa, H. (2012). SOCS, 594

inflammation and autoimmunity. [Review]. Frontiers in Immunology, 3. doi: 595

10.3389/fimmu.2012.00020 596

Zheng, S. G. (2013). Regulatory T cells vs Th17: differentiation of Th17 versus Treg, are the 597

mutually exclusive? Am J Clin Exp Immunol, 2(1), 94-106. 598

Zhou, L., Chong, M. M., & Littman, D. R. (2009). Plasticity of CD4+ T Cell Lineage 599

Differentiation. Immunity, 30(5), 646-655. 600

Zhou, L., Ivanov, I. I., Spolski, R., Min, R., Shenderov, K., Egawa, T., Littman, D. R. (2007). 601

IL-6 programs TH-17 cell differentiation by promoting sequential engagement of the 602

IL-21 and IL-23 pathways. Nature immunology, 8(9), 967-974. 603

Zhou, L., Lopes, J. E., Chong, M. M., Ivanov, II, Min, R., Victora, G. D., Littman, D. R. 604

(2008). TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing 605

RORgammat function. Nature, 453(7192), 236-240. doi: 10.1038/nature06878 606

Zhu, J., Yamane, H., & Paul, W. E. (2010). Differentiation of effector CD4 T cell populations 607

(*). Annu Rev Immunol, 28, 445-489. doi: 10.1146/annurev-immunol-030409-101212 608

Ziegler, S. F., & Buckner, J. H. (2009). FOXP3 and the regulation of Treg/Th17 609

differentiation. Microbes Infect, 11(5), 594-598. doi: 10.1016/j.micinf.2009.04.002 610

(27)

27 List of figures:

612

Fig. 1. 613

MTS assay examining the effects of SCFAs on the viability and proliferation of PBMCs. 614

Cells were sub-cultured in 96 well plates at initial density of 103 cells per 100 μL with or 615

without SCFAs and/or LPS and incubated for 48 h. PBMCs in growth media was set as 616

control. Cells viability (%) = (OD of SCFAs treated sample / OD of control sample) ×100. 617

Ace = acetate, Pro = propionate, But = Butyrate, PBMCs = Peripheral blood monocular cells. 618

Results are expressed as mean of five independent experiments with error bars showing the 619

standard deviation, (p < 0.05). 620

621

Fig. 2. 622

Nitric oxide (NO) concentration (µM) in supernatants of stimulated PBMCs (1× 106 cells/ 623

mL) cultures. PBMCs were sub-cultured for 48 h either in the presence of LPS (5 μg/mL), 624

LPS with each SCFA (acteate, propionate or butyrate) or LPS with the mixed SCFAs. RPMI 625

media only was used as a control. Supernatants were collected after 24 h and 48 h and the 626

level of NO was determined by the Griess reaction. NO levels were significantly different (p 627

< 0.05) from the control values (bars with different letter). NO = Nitric oxide; LPS = 628

Lipopolysaccharide; Ace = acetate; Pro = propionate; But = butyrate; SCFAs = short chain 629

fatty acids. Data represent the mean of five independent experiments with error bars showing 630

the standard deviation. 631

632

Fig. 3. 633

Cytokine concentrations of IL-1β (A), IL-2 (B), IL-6 (C), IL-10 (D), IL-17 (E), TGF-β1 (F), 634

IL-21 (G) and IL-23 (H) in supernatant after PBMCs (1 × 106 cells/mL) were sub-cultured 635

with either acetate, propionate, butyrate or mixed SCFAs for 24 h and 5 μg/mL. LPS was 636

added to each well and incubated for a further 24 h or 48 h. PBMCs with LPS only was set as 637

control. Data represent the mean of five independent experiments with error bars showing the 638

standard deviation, (bars with different letter are significantly different (p < 0.05)). SCFAs = 639

short chain fatty acids; IL = interleukin; TGF = transforming growth factor; Ctrl = control, 640

LPS = Lipopolysaccharide, Ace = acetate, Pro = propionate, But = butyrate, Mix = Mixed 641 SCFAs. 642 643 Fig. 4. 644

Expression of activation marker CD25 on T lymphocytes, plots were gated on CD3 (A), 645

percentage of induced CD25+ forkhead box protein 3 (FoxP3+) (B), and induction of ROR-γt 646

expressing T helper type 17 (Th17) (C) by PBMCs in response to LPS and/or SCFAs. 647

(28)

28 Table 1

650

Cytokine levels (pg/mL) produced by PBMCs co-cultured with LPS only for 24 h then SCFAs were added and the cells were incubated for 651

another 24 h. 652

Cytokines T(h) Ctrl LPS Ace Pro But Mix

IL-β1 24 55.04 ±6.3 1587.1 ±203.8 1571.7 ±130.2 1275.2 ±51.2 1601.1± 105.4 1620.1 ±33.0 48 53.13 ±5.8 1828.2 ±166.4 1031.7 ±35.5 1121.2 ±48.4 992.5 ±166.3 1086.3 ±143.0

IL-2 24 17.33 ±0.5 285.13 ±5.1 291.32 ±12.2 287.23 ±8.5 290.1 ±41.7 289.21 ±6.1

48 15.46 ±1.3 346.21 ±21.1 225 ±31.5 234 ±19.8 201.3 ±32.2 222 ±7.6

IL-6 24 59.36 ±3.3 1082.07 ±20.6 1001.21 ±25.7 1013 ±5.1 1014.12 ±83.3 997.39 ±64.6

48 71.41 ±5.2 1097.13 ±11.9 831.3 ±32.4 942 ±25.1 612 ±32.9 741 ±46.2

IL-10 24 71.12 ±4.2 76.18 ±3.6 78.3 ±13.3 88.1 ±1.2 87.23 ±4.8 76.91 ±3.5

48 77.34 ±3.2 74.75 ±2.8 181.2 ±6.4 173.15 ±8.1 26 ±12 263 ±20.8

IL-17 24 74.13 ±5.3 403.17 ±34.1 423.1 ±8.7 411.1 ±21.1 435.2 ±23.0 429.19 ±46.4

48 78.41 ±1.9 432 ±14.1 107.4 ±2.8 172 ±5.3 121 ±10.5 133 ±16.8

IL-21 24 231.12 ±14.4 371.41 ±6.9 372.42 ±95.9 358 ±18.5 364.3 ±28.0 361.1 ±17.1

48 236.71 ±12 391.35 ±19.9 62.21 ±3.3 117.7 ±5.9 61.5 ±9.4 75.13 ±4.0

IL-23 24 81.28 ±3.8 1271 ±89.8 1237 ±175.1 1211.5 ±81.1 1301 ±178.4 1291 ±38.3

48 86.30 ±3.2 1295 ±116.0 920 ±34.1 989 ±32.1 931.2 ±11.7 1011 ±26.8

TGF-β1 24 161.1 ±9.1 1751 ±127.5 1723 ±108.2 1715 ±81.1 1695.4 ±17.7 1699 ±116.5

48 156.3±4.1 1950±38.4 1710±46.1 1608±135.6 1651±31.5 1558±174.0

IL = interleukin; TGF-β1 = transforming growth factor beta 1; Ctrl = control (PBMCs without stimulation), LPS = Lipopolysaccharide, Ace = 653

(29)

29 Fig. 1.

655

656 657

92 93 94 95 96 97 98 99 100 101

PBMCs Ace Pro But SCFAs

C

ells

V

ia

b

ilit

y

%

SCFAs (2mM)

LPS

(30)

30 Fig. 2.

658

659

0 10 20 30 40 50 60 70 80 90

Control LPS Act Pro But SCFAs

µ

M

o

f N

O

SCFA 2 mM

0 24 48

a a a a a a a a

b b

c c

c c c c

a a a a a

b b

(31)

31 Fig. 3.

660

661

662

0 400 800 1200 1600 2000

1 1.5 2 1 1.5 2 1 1.5 2 1 1.5 2

Ctrl LPS Acetate Propionate Butyrate Mixed

IL

-1

β

co

n

cen

tra

ti

o

n

n

g

/m

L

24 48

A

a b

c c

b bc

c c c c c c c

c

0 100 200 300 400

1 1.5 2 1 1.5 2 1 1.5 2 1 1.5 2

Ctrl LPS Acetate Propionate Butyrate Mixed

IL

-2 c

on

ce

n

tr

at

ion

p

g/

m

L

24 48

B

a b

c c c c c c

(32)

32 663

664

665

0 200 400 600 800 1000 1200 1400

1 1.5 2 1 1.5 2 1 1.5 2 1 1.5 2

Ctrl LPS Acetate Propionate Butyrate Mixed

IL

-6 c

on

ce

n

tr

at

ion

p

g/

m

L

24 48

C

a b

cd

c c c c c c

d d c

cd cd

0 50 100 150 200 250 300

1 1.5 2 1 1.5 2 1 1.5 2 1 1.5 2

Ctrl LPS Acetate Propionate Butyrate Mixed

IL

-10 c

on

ce

n

tr

at

ion

p

g/

m

L

24 48

D

a a

b b b b b b

d d d d

(33)

33 666

667

668

0 100 200 300 400 500

1 1.5 2 1 1.5 2 1 1.5 2 1 1.5 2

Ctrl LPS Acetate Propionate Butyrate Mixed

IL

-17 c

on

ce

n

tr

at

ion

p

g/

m

L

24 48

E

a b

a a c

c c

c c c c

ac ac

ac

0 400 800 1200 1600 2000

1 1.5 2 1 1.5 2 1 1.5 2 1 1.5 2

Ctrl LPS Acetate Propionate Butyrate Mixed

T

GF

1

con

ce

n

tr

at

ion

p

g/

m

L

24 48

F

a b

b b b

(34)

34 669

670

671

0 100 200 300 400 500

1 1.5 2 1 1.5 2 1 1.5 2 1 1.5 2

Ctrl LPS Acetate Propionate Butyrate Mixed

IL

-21 c

on

ce

n

tr

at

ion

p

g/

m

L

24 48

G

a b

c c c c c c c c c d d d

0 300 600 900 1200 1500

1 1.5 2 1 1.5 2 1 1.5 2 1 1.5 2

Ctrl LPS Acetate Propionate Butyrate Mixed

IL

-23 c

on

ce

n

tr

at

ion

p

g/

m

L

24 48

H

a b

(35)

35 Fig. 4.

(36)
(37)

Figure

Table 1  Cytokine levels (pg/mL) produced by PBMCs co-cultured with LPS only for 24 h then SCFAs were added and the cells were incubated for
Fig. 2.
Fig. 3. 2000

References

Related documents

If signals are reserved for truly exceptional conditions it may be better to code for readability. The equivalent code fragment in 4.4 is more readable than this

Among uninsured children who lived in rural areas, the average distance to the nearest federally funded community health center, public housing primary care program, or migrant

Maxima ofthe enzyme activity were identical to that ofMDH to a considerable extent and both enzymes had approximately similar activity at over-ripe stage whereas their

The probation officer inter- viewed the patient once a month and psychiatric sessions were arranged twice weekly for the first year, and once a week during the

It has been known for a long time that ion clusters in this size range exist in the atmosphere, but the authors demonstrate that an ion-induced nucleation process by activation of

In [6] proposed two main clustering based segmentation methods named Fuzzy C-Means and K-Means to detect brain tumor.. By changing the image mode to gray-scale, remove the

The framework presented in figure 3 aims at introducing a systematic approach to dealing with administrative issues of pollution connecting ports authority, other

(B) DLEC1 protein expression was detected by Western blot in human normal tissues of esophageal, lung and testis, immortalized esophageal epithelial cell lines and